CN112569242A - Novel treatment for cancer - Google Patents

Novel treatment for cancer Download PDF

Info

Publication number
CN112569242A
CN112569242A CN202011468330.5A CN202011468330A CN112569242A CN 112569242 A CN112569242 A CN 112569242A CN 202011468330 A CN202011468330 A CN 202011468330A CN 112569242 A CN112569242 A CN 112569242A
Authority
CN
China
Prior art keywords
cancer
pyridin
diazepan
piperidin
carboxamide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202011468330.5A
Other languages
Chinese (zh)
Inventor
彼得·理查森
杰奎琳·玛丽·沃林
克劳迪奥·费斯图恰
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
BenevolentAI Cambridge Ltd
Original Assignee
Proximagen Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Proximagen Ltd filed Critical Proximagen Ltd
Publication of CN112569242A publication Critical patent/CN112569242A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • A61K31/55131,4-Benzodiazepines, e.g. diazepam or clozapine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Landscapes

  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Dermatology (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present application relates to novel treatments for cancer. The present application relates to 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide, or a pharmaceutically acceptable salt thereof, for use in the treatment of CNS cancer. The application also relates to combination therapy with radiation and/or chemotherapeutic agents for the treatment of cancer.

Description

Novel treatment for cancer
The application is a divisional application of Chinese patent application 201680020617.7, and the original application is an application entering the Chinese national stage at 09 and 30 months in 2017 according to the international application number PCT/IB2016/051880 and the international application date 2016, 04 and 01 days in 2016.
Technical Field
The present application relates to the field of cancer therapy. The invention described herein relates to the use of the CXCR4 antagonist 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide in the treatment of CNS cancer. The invention also relates to the use of 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide in combination with radiation and/or chemotherapeutic agents for the treatment of cancer, including CNS cancer.
Background
CXCR4 is a G protein-coupled receptor whose natural endogenous ligand is the cytokine SDF-1 (stromal derived factor-1); also known as CXCL 12). CXCR4 was originally discovered as a co-receptor for CD4, which was used to bring (X4) HIV-1 of the T-cell lineage into T cells. Control of CXCR4 (manipulation), in combination with granulocyte colony-stimulating factor (G-CSF), has been demonstrated to improve the results of hematopoiesis (broxmeye et al, 2005) and stem cell mobilization (mobilization) of endothelial progenitor cells (Pitchford et al, 2009). CXCR4-SDF-1 interaction is also a major regulator of cancer stem cell trafficking in humans (Croker and alan, 2008), and plays a key role in the progression and metastasis of multiple types of cancer cells in organs that highly express SDF-1 (Zlotnik, 2008).
Several types of cancer (including non-small cell lung cancer, breast cancer and glioblastoma) express CXCR4 and SDF-1, which are closely associated with the maintenance of cancer stem cells (Wang et al, 2006; Croker and alan, 2008) and the recurrence of tumors after treatment. Furthermore, CXCR4 has been shown to play a role in the formation of new blood vessels in experimental tumors (Kioi et al, 2010).
Of particular interest, it was observed that CXCR4 expression in many cancers is associated with small cell populations that exhibit stem cell-like characteristics (i.e., they are tumorigenic). These stem cell-like cells are enriched under specific tissue culture conditions (serum-free plus EGF and FGF) and are closely related to mediating metastatic spread (see, e.g., Hermann et al, 2007). In CNS cancers, including primary brain tumors, these cells are closely associated with the spread of the cancer through the brain (Zagzag et al, 2008).
In humans, CNS cancers include gliomas, the most common primary brain tumor type. Gliomas originate from the supporting glial cells (supporting glia cells) of the brain and are often associated with a severe prognosis. Based on the source cells, gliomas include: astrocytomas, ependymomas, oligodendrogliomas (oligodendrogliomas), glioblastoma, oligodendroglioma, and the like. Higher astrocytomas, including glioblastoma multiforme (GBM) and Anaplastic Astrocytomas (AA), are the most common intrinsic brain tumors in adults.
Gliomas are defined histologically by whether they exhibit predominantly astrocytic or oligodendroglial cell morphology. Gliomas are classified by cellular, nuclear atypia, necrosis, mitotic features and microvascular hyperplasia-all features associated with the behavior of biological invasiveness. This diagnostic system has been developed in decades of clinical experience with gliomas and is now the cornerstone of neurooncology. The classification scheme for astrocytomas by the world health organization is classified into four (4) grades. The less malignant tumors belong to grade I (fibroid astrocytoma) and II (astrocytoma), while the more malignant tumors are designated grade III (anaplastic astrocytoma) and grade IV (GBM). Oligodendrogliomas and mixed gliomas (gliomas with both oligodendrocyte and astrocytic components) are present in both the lower (grade II) and more malignant variants (grade III).
These tumors are usually treated in primary diagnosis with a combination of surgery, focused irradiation, and the DNA alkylating agent temozolomide. However, in some patients, the tumor regrows, indicating that the tumor is or has become resistant to temozolomide. Resistance to temozolomide is typically the result of expression of the DNA repair enzyme O-6-methylguanine-DNA methyltransferase (MGMT). Metastatic cancers of the CNS (i.e., those arising from the spread of peripheral cancers such as breast and lung cancer) are treated in a similar manner, but sometimes with whole brain rather than focused irradiation. It is not always possible or desirable to treat CNS cancers by surgery, for example, tumors may be inaccessible (e.g., deep in the brain), or patients may not tolerate the trauma of neurosurgery (possibly because they are elderly and/or infirm). Irradiation (radiotherapy) and treatment with cytotoxic agents (chemotherapy) are known to have undesirable side effects. Thus, there is an unmet medical need for the treatment of CNS cancers, including brain cancers. Very few chemotherapeutic agents penetrate sufficiently into the brain to achieve effective therapeutic concentrations there, which makes it difficult to treat CNS cancers with systemically administered chemotherapeutic agents. One agent that does enter the brain is lomustine, a DNA alkylating agent that has been widely used in clinical trials for brain cancer. Other agents include temozolomide, carmustine, irinotecan, and carboplatin.
Studies have reported the treatment of CNS cancers in mice using the CXCR4 antagonist AMD3100 in combination with irradiation or chemotherapeutic agents (e.g., Redjal et al, 2006; and Chen et al, 2013). However, it is expected that patients treated with a combination of AMD3100 and a radiation and/or chemotherapeutic agent will suffer greater toxic side effects than patients treated with AMD3100 or a radiation and/or chemotherapeutic agent alone. Bone marrow is known to provide a protective and nutritional environment for Hematopoietic Stem Cells (HSCs), which are essential for maintaining a blood cell supply. Treatment with CXCR4 antagonists (e.g., AMD3100) can mobilize HSCs from the bone marrow. When administered with GCSF, it mobilizes enough HSCs to allow HSC transplantation (i.e., HSCs are collected and stored prior to administration of HSCs to patients who have undergone aggressive chemotherapy). The method is particularly useful for treating bone marrow cancer (e.g., multiple myeloma) as it allows aggressive chemotherapy (aggressive chemotherapy) followed by bone marrow recovery (Di Persio et al, 2009; Micallef et al, 2009). The cytoprotective properties of bone marrow are seen in the case of HSCs (Kopp et al, 2005) as well as some cancer stem cells (such as those of acute lymphoblastic leukemia) (Colmone et al, 2008; Yang et al, 2013).
Patients treated with chemotherapy and/or radiation therapy often experience side effects due to the destruction of bone marrow HSCs. It is expected that release of HSCs from the protective environment of the bone marrow will exacerbate these side effects, possibly leading to anemia and neutropenia. Thus, there is an unmet medical need for a combination of CXCR4 antagonists and chemotherapeutic agents for the treatment of cancer (including CNS cancers), with reduced risk of side effects.
CXCR4 antagonists are known in the literature. For example, WO2012/049277 teaches the structure and preparation of the CXCR4 antagonist 6- {4- [1- (prop-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide, which is example 30 and has the following structure:
Figure BDA0002833537670000031
disclosure of Invention
In a first aspect of the invention, the applicants have found that 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide is surprisingly effective in the treatment of CNS cancers, including brain cancers, also known as orthotopic (intracranial) tumors.
In a second aspect of the invention, applicants have found that the CXCR4 antagonist 6- {4- [1- (prop-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide in combination with radiation and/or chemotherapeutic agents is surprisingly effective (i.e. synergistic) in treating cancer, including CNS cancer.
With respect to the second aspect of the present invention, the applicant has additionally found that treatment with 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide in combination with radiation and/or chemotherapeutic agents unexpectedly reduces the risk of side effects in patients. In other words, the present invention enables a combination therapy for cancer comprising 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide with unexpectedly improved safety and irradiation and/or chemotherapeutic agents.
Drawings
FIG. 1 is a graph showing the degree of mobilization of Hematopoietic Stem Cells (HSCs) and progenitor cells (CFU-GEMM) in mice following injection of vehicle AMD3100(5mg/kg) and 6- {4- [1- (prop-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide (30 mg/kg).
FIG. 2 is a graph showing that treatment with 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide (dashed line) inhibited the growth of human glioblastoma cell line (T98G) in nude mouse subcutaneous xenografts compared to control (solid line).
FIG. 3 is a graph showing that treatment with 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide with the chemotherapeutic agent temozolomide inhibited the growth of a human glioblastoma cell line (T98G) in subcutaneous xenografts in nude mice. The combination of the two treatments resulted in unexpectedly improved (i.e., synergistic) antitumor efficacy.
FIG. 4 is a graph showing that treatment with 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide with the chemotherapeutic agent bevacizumab inhibits the growth of tumors formed by a human glioblastoma cell line (U87MG) introduced intracranially in nude mice. The combination of the two treatments resulted in an unexpectedly improved (i.e. synergistic) antitumor efficacy as evidenced by the prolonged duration of survival of mice with in situ (intracranial) tumors (combination p ═ 0.002, HR 3.4 versus vehicle).
FIG. 5 is a graph showing that treatment with 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide with the chemotherapeutic agent temozolomide inhibited the growth of tumors formed by the human glioblastoma cell line (U87MG) introduced intracranially in nude mice. The combination of the two treatments resulted in an unexpectedly improved (i.e. synergistic) antitumor efficacy as demonstrated by the prolonged duration of survival of mice with in situ (intracranial) tumors (combination p ═ 0.02, HR 2.8 versus temozolomide alone).
FIG. 6 is a graph showing that treatment with 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide with radiation inhibits the growth of tumors formed by transcranially introduced human glioblastoma cell line (U87MG) in nude mice. The combination of the two treatments resulted in an unexpectedly improved (i.e. synergistic) antitumor efficacy as evidenced by the prolonged duration of survival of mice with in situ (intracranial) tumors (combination p ═ 0.0002, HR 4.0 versus radiotherapy alone).
Figure 7 is a graph showing that treatment with 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide with sunitinib inhibited the growth of tumors formed by a human glioblastoma cell line (U87MG) introduced intracranially in nude mice. The combination of the two treatments resulted in an unexpectedly improved (i.e. synergistic) antitumor efficacy as demonstrated by the prolonged duration of survival of mice with in situ (intracranial) tumors (combination p ═ 0.2, HR 1.6 versus vehicle).
FIGS. 8A and 8B are graphs showing that treatment with 6- {4- [1- (prop-2-yl) piperidin-4-yl ] -1, 4-. diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide delays the growth of tumors formed by a human glioblastoma cell line (U87MG) introduced intracranially in nude mice, and 6- {4- [1- (prop-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide exerts a synergistic effect with bevacizumab or sunitinib in delaying or inhibiting the growth of tumors. The arrow on the X-axis indicates the end of dosing.
Figures 9A and 9B are graphs showing that treatment with 6- {4- [1- (prop-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide delays intracranial tumor growth and exerts a synergistic effect with radiation therapy (figure 9A) and temozolomide treatment (figure 9B) in delaying or inhibiting the growth of tumors formed by a human glioblastoma cell line (U87MG) introduced intracranially into nude mice. The arrow on the X-axis indicates the end of the dose.
Figure 10 is a graph showing that treatment with 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide unexpectedly improved the efficacy of combined temozolomide and radiation treatment in survival of mice with in situ (intracranial) tumors. The arrow on the X-axis indicates the end of the dose.
Detailed Description
In one embodiment according to the first aspect of the invention, applicants have made available 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide, or a pharmaceutically acceptable salt thereof, for use in the treatment of CNS cancer. In one embodiment, the CNS cancer is a brain cancer. In one embodiment, the CNS cancer is glioma. In one embodiment, the CNS cancer is selected from: neuroblastoma, glioblastoma, other astrocytoma, oligodendroglioma, meningioma, ependymoma, oligodendroglioma, medulloblastoma and metastatic cancer that enters the CNS from peripheral cancers. In one embodiment, the CNS cancer is selected from the group consisting of glioblastoma and astrocytoma.
In one embodiment according to the first aspect of the present invention, applicants have made available the use of 6- {4- [1- (prop-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl) -N- (pyridin-4-yl) pyridine-2-carboxamide, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of CNS cancer. In one embodiment, the CNS cancer is a brain cancer. In one embodiment, the CNS cancer is glioma. In one embodiment, the CNS cancer is selected from: neuroblastoma, glioblastoma, other astrocytoma, oligodendroglioma, meningioma, ependymoma, oligodendroglioma, medulloblastoma and metastatic cancer that enters the CNS from peripheral cancers. In one embodiment, the CNS cancer is selected from the group consisting of glioblastoma and astrocytoma.
In one embodiment according to the first aspect of the present invention, applicants make available a method of treating a patient suffering from a CNS cancer, said method comprising administering to the patient 6- {4- [ l- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide, or a pharmaceutically acceptable salt thereof, in an amount sufficient to provide a therapeutic effect. In one embodiment, the CNS cancer is a brain cancer. In one embodiment, the CNS cancer is glioma. In one embodiment, the CNS cancer is selected from: neuroblastoma, glioblastoma, other astrocytoma, oligodendroglioma, meningioma, ependymoma, oligodendroglioma, medulloblastoma and metastatic cancer that enters the CNS from peripheral cancers. In one embodiment, the CNS cancer is selected from the group consisting of glioblastoma and astrocytoma.
In one embodiment according to the second aspect of the present invention, applicants have made available 6- {4- [1- (prop-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide in combination with irradiation for the treatment of cancer.
In another embodiment according to the second aspect of the invention, applicants have made it feasible that 6- {4- [1- (prop-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide is used in combination with one or more chemotherapeutic agents, including brain penetrating chemotherapeutic agents, for the treatment of cancer.
In another embodiment according to the second aspect of the invention, applicants make feasible the combination of 6- {4- [1- (prop-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide with one or more chemotherapeutic agents for the treatment of cancer.
Without wishing to be bound by theory, it is understood that the reduced risk of side effects after administration of the combination according to the second aspect of the invention is due to the unexpectedly low tendency of 6- {4- [1- (prop-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide to mobilize Hematopoietic Stem Cells (HSCs) from the protective environment of the bone marrow. The advantage of this reduced mobilization is that during treatment with 6- {4- [1- (prop-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide in combination with irradiation and/or chemotherapeutic agents, HSCs tend to remain in the protective environment of the bone marrow and are therefore less likely to be damaged by irradiation and/or chemotherapeutic agents. This results in a reduced likelihood of side effects due to HSC destruction and subsequent cytopenia (e.g., anemia and neutropenia).
In one embodiment of the compound, use or method according to the second aspect of the present invention, the cancer comprises the following cancers and metastases thereof: cancers of the lung (including non-small cells and small cells), pancreas, cervix, thyroid, kidney, ovary, prostate, skin (including melanoma); cancers of the GI tract (including esophagus, liver, colorectal, and stomach); squamous cell carcinoma of the oral cavity; hematological cancers including leukemias such as B-CLL, AML, CML, ALL; lymphomas such as intraocular, non-hodgkin and hodgkin lymphomas, and multiple myeloma; cancers of the nervous system, including brain cancer, neuroblastoma, glioblastoma, other astrocytoma, oligodendroglioma, meningioma, ependymoma, oligodendroglioma, medulloblastoma, and metastatic cancers that enter the CNS from peripheral cancers.
In one embodiment of the compound, use or method according to the second aspect of the present invention, said cancer is selected from CNS cancers of: neuroblastoma, glioblastoma, other astrocytoma, oligodendroglioma, meningioma, ependymoma, oligodendroglioma, medulloblastoma and metastatic cancer that enters the CNS from peripheral cancers. In one embodiment, the CNS cancer is selected from the group consisting of glioblastoma and astrocytoma.
In one embodiment of the compound, use or method according to the second aspect of the invention, the chemotherapeutic agent is a DNA modifying agent.
In one embodiment of the compound, use or method according to the second aspect of the invention, the chemotherapeutic agent (e.g. temozolomide) is detrimental or otherwise toxic to hematopoietic stem cells.
In one embodiment of the compound, use or method according to the second aspect of the invention, 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide is combined with a chemotherapeutic agent selected from the group consisting of: bevacizumab, sunitinib, temozolomide, vincristine, lomustine, procarbazine, carmustine, irinotecan, cisplatin, carboplatin, methotrexate, etoposide, bleomycin, vinblastine, actinomycin D, cyclophosphamide and ifosfamide. In a preferred embodiment, the chemotherapeutic agent is bevacizumab. In a preferred embodiment, the chemotherapeutic agent is sunitinib. In a preferred embodiment, the chemotherapeutic agent is temozolomide. In a preferred embodiment, the chemotherapeutic agent is vincristine. In a preferred embodiment, the chemotherapeutic agent is lomustine. In a preferred embodiment, the chemotherapeutic agent is procarbazine. In a preferred embodiment, the chemotherapeutic agent is carmustine. In a preferred embodiment, the chemotherapeutic agent is irinotecan. In a preferred embodiment, the chemotherapeutic agent is cisplatin. In a preferred embodiment, the chemotherapeutic agent is carboplatin. In a preferred embodiment, the chemotherapeutic agent is methotrexate. In a preferred embodiment, the chemotherapeutic agent is etoposide. In a preferred embodiment, the chemotherapeutic agent is bleomycin. In a preferred embodiment, the chemotherapeutic agent is vinblastine. In a preferred embodiment, the chemotherapeutic agent is actinomycin D. In a preferred embodiment, the chemotherapeutic agent is cyclophosphamide. In a preferred embodiment, the chemotherapeutic agent is ifosfamide.
In one embodiment of the compound, use or method according to the second aspect of the present invention, the chemotherapeutic agent is capable of penetrating the brain and reaching a therapeutic concentration therein after systemic administration to a patient. In one embodiment of the compound, use or method according to the second aspect of the present invention, the brain penetrating chemotherapeutic agent is selected from any one of sunitinib, lomustine, temozolomide, carmustine, irinotecan and carboplatin. In one embodiment, the brain penetrating chemotherapeutic agent is lomustine or carmustine.
In one embodiment of the compound, use or method according to the second aspect of the invention, 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide is administered prior to administration of the radiation and/or chemotherapeutic agent.
In one embodiment of the compound, use or method according to the second aspect of the invention, 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide is administered concurrently with the administration of the radiation and/or chemotherapeutic agent.
In one embodiment of the compound, use or method according to the second aspect of the invention, 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide is administered after administration of the radiation and/or chemotherapeutic agent.
In one embodiment of the compound, use or method according to the second aspect of the present invention, the cancer to be treated comprises a tumor that is resistant to temozolomide. In one embodiment of the compound, use or method according to the second aspect of the invention, the cancer to be treated comprises a tumour that is resistant to irradiation. In one embodiment of the compound, use or method according to the second aspect of the present invention, the cancer to be treated comprises a tumor that is resistant to temozolomide and radiation.
In one embodiment of the compound, use or method according to the second aspect of the invention, 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide is in an intravenous formulation.
In one embodiment of the compound, use or method according to the second aspect of the invention, the chemotherapeutic agent is in an intravenous formulation.
In another embodiment, 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide is used in combination with a chemotherapeutic agent capable of penetrating the blood-brain barrier.
In one embodiment, 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide is used in combination with external beam (external beam) radiation therapy 60Gy in the 2Gy section.
In one embodiment, 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide is used in combination with external beam radiation therapy 60Gy in fraction 2Gy and temozolomide.
It is expected that the claimed combinations will be particularly effective in treating cancers that have become resistant or otherwise unresponsive to treatment with temozolomide and/or radiation.
Term(s) for
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are now described.
As used herein and in the appended claims, the term "a" means "an" or "an" unless the context clearly dictates otherwise. Thus, for example, reference to "a method" includes one or more methods and/or steps of the type described herein, which will become apparent to those skilled in the art upon reading this disclosure and so forth.
The terms "treating cancer" and "treating CNS cancer" as used herein are not meant to be absolute terms. In some aspects, the compositions and methods of the invention are directed to reducing the size of a tumor or the number of cancer cells, resulting in remission of the cancer, inhibiting or preventing an increase in the size of a tumor or the number of cells of cancer cells. In certain instances, treatment with a compound or combination according to the claimed invention results in an improved prognosis. Treatment as a prophylactic measure (i.e., prophylaxis) is also included. For example, a patient at risk of developing or reoccurring cancer may be treated as described herein.
The term "cancer" as used herein refers to a broad type of disorder characterized by hyperproliferative cell growth in vitro (e.g., transformed cells) or in vivo. Conditions that may be treated or prevented by the compositions and methods of the present invention include, for example, a variety of tumors, including benign or malignant tumors, various hyperplasias, and the like. The compounds and methods of the first and second aspects of the invention may effect inhibition and/or reversal of undesired hyperproliferative cell growth associated with such conditions. The term "cancer" includes any solid tumor or liquid cancer (liquid cancer), and may be metastatic or non-metastatic. Examples of cancers and their metastatic cancers that are susceptible to treatment with the claimed compounds or combinations include Central Nervous System (CNS) cancers.
The term "cancer of the CNS" as used herein includes brain cancers such as gliomas, neuroblastoma, glioblastoma, other astrocytomas, oligodendroglioma, meningioma, ependymoma and medulloblastoma. Gliomas are tumors that arise from glial cells or their precursors of the brain or spinal cord. Gliomas are histologically defined based on whether they exhibit predominantly astrocytic or oligodendrocyte morphology and are graded by cellular, nuclear atypic, necrotic, mitotic features and microvascular proliferation-all features associated with bioinvulsive behavior. There are two major types of astrocytomas-high and low. Higher tumors grow rapidly, vascularize well, and can readily diffuse through the brain. Low-grade astrocytomas are usually localized and grow slowly over a long period of time. Advanced tumors are more aggressive, require very high intensity treatment, and are associated with shorter survival time lengths compared to low-grade tumors. Most astrocytic tumors are low grade in children and high grade in adults. These tumors can occur anywhere in the brain and spinal cord. Some of the more common low-grade astrocytomas are: juvenile hairy cell Astrocytoma (JPA), fibroastrocytoma Pleomorphic Astrocytoma (PXA) and dysplastic Neuroepithelial tumors (DNET). The two most common higher astrocytomas are Anaplastic Astrocytomas (AA) and glioblastoma multiforme (GBM).
Further examples of cancers and their metastatic cancers that are sensitive to treatment with the claimed combination include cancers of the lung (including non-small cells and minicells), pancreas, cervix, thyroid, kidney, ovary, prostate, skin (including melanoma), cancers of the GI tract (including esophagus, liver, colorectal and stomach), oral squamous cell carcinoma, hematological cancers including leukemias such as B-CLL, AML, CML, ALL, lymphomas such as intraocular, non-hodgkin and hodgkin lymphomas, and multiple myeloma.
The term "patient with cancer" as used herein refers to an individual or subject who has been diagnosed with cancer or a cell proliferative disorder.
The term "patient with CNS cancer" as used herein refers to an individual or subject who has been diagnosed with CNS cancer or a cell proliferative disorder of the CNS (including brain cancer) and an in situ (intracranial) tumor.
The term "chemotherapeutic agent" as used herein is any anti-cancer drug or drug having anti-cancer cell activity. Chemotherapeutic agents include monoclonal antibodies and small molecule drugs. Some small molecule chemotherapeutic drugs are cytotoxic, that is, they act by killing rapidly dividing cells. Examples of chemotherapeutic agents include bevacizumab, sunitinib, temozolomide, vincristine, lomustine, procarbazine, carmustine, irinotecan, cisplatin, carboplatin, methotrexate, etoposide, bleomycin, vinblastine, actinomycin D, cyclophosphamide, and ifosfamide. Chemotherapeutic drugs can be administered one drug at a time (single agent chemotherapy) or in combination (combination chemotherapy). Chemotherapeutic drugs may be administered in combination with irradiation. In one embodiment, the chemotherapeutic agent is other than 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide. In one embodiment, the chemotherapeutic agent is an antibody, such as bevacizumab. In one embodiment, the chemotherapeutic agent is sunitinib.
Any suitable amount and type of radiation and/or chemotherapeutic agent may be used in combination with 6- {4- [1- (prop-2-yl) piperidin-4-yl ] -l, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide in the present invention. Examples of suitable irradiation regimens and chemotherapeutic agents can be found in the Current guidelines 2011Canada, Easaw et al, Current Oncology, vol 18, phase 3.
The term "brain-penetrating chemotherapeutic agent" as used herein means a chemotherapeutic agent that, when administered systemically, is capable of penetrating into the brain and reaching effective therapeutic concentrations therein. Examples of the brain penetrating chemotherapeutic agent include sunitinib, lomustine, temozolomide, carmustine, irinotecan, and carboplatin.
The term "therapeutic effect" as used herein means providing a therapeutic response in a subject. For example, a therapeutic effect comprising inhibiting tumor progression or tumor growth is provided. One skilled in the art will appreciate that tumor progression in a human patient can be determined by a variety of methods. For example, the size of a tumor near the skin can be measured by determining the width and depth of the tumor with a caliper (caliper) and then calculating the tumor volume. Tumors that are not readily accessible, such as lung and CNS cancers, can be measured by observing images obtained from Magnetic Resonance Imaging (MRI) scans. CNS tumors (e.g., brain tumors) can be measured by a combination of MRI scanning and by monitoring nervous system performance. Growth of brain tumors is often associated with reduced nervous system performance. Providing a therapeutic effect also includes extending the survival of the patient or subject beyond that expected in the absence of treatment. In one embodiment, treatment of a patient or subject with a compound or combination according to the first or second aspect of the invention extends survival to 1 month or several months, preferably 3 months or more, more preferably 6 months or more, more preferably 1 year or more, preferably 2 years or more, or 3 years or more, even preferably 5 years or more, including 10 years or more, beyond what would be expected in the absence of treatment. Providing a therapeutic effect also includes eliminating cancer cells. Providing a therapeutic effect also includes a reduction in tumor volume.
The term "irradiation" as used herein includes any suitable type and amount of irradiation that provides a therapeutic effect. Examples of suitable irradiation regimens and chemotherapeutic agents can be found in the Current guidelines 2011Canada, Easaw et al, Current Oncology, vol 18, phase 3.
The term "salt" as used herein includes base addition, acid addition and ammonium salts. 6- {4- [1- (Prop-2-yl) piperidin-4-yl]-1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide is basic and thus formsSalts, including pharmaceutically acceptable salts formed with inorganic acids (e.g., with hydrohalic acids such as hydrochloric or hydrobromic, sulfuric, nitric or phosphoric acids, and the like) and with organic acids (e.g., with acetic, trifluoroacetic, tartaric, succinic, fumaric, maleic, malic, salicylic, citric, methanesulfonic, p-toluenesulfonic, benzoic, benzenesulfonic, glutamic, lactic and mandelic acids, and the like). Those compounds having a basic nitrogen may also form quaternary ammonium salts with pharmaceutically acceptable counterions such as chloride, bromide, acetate, formate, p-toluenesulfonate, succinate, hemisuccinate, naphthalene-disulfonate, methanesulfonate, trifluoroacetate and the like. For a review of salts, see Stahl and Wermuth“Handbook of Pharmaceutical Salts:Properties,Selection and Use”(Wiley-VCH,Weinheim,Germany,2002)。
The compound "6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide" may be present as a solvate. The term "solvate" is used herein to describe a molecular complex comprising a compound of the invention and a stoichiometric amount of one or more pharmaceutically acceptable solvent molecules (e.g., ethanol). When the solvent is water, the term "hydrate" is used.
The compound "6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide" may exist in amorphous form and/or in several polymorphic forms and may be obtained in different crystal habits. Any reference herein to 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide includes all forms of the compound, whether amorphous or polymorphic.
Pharmaceutical formulations and preparations
The 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide used in the present invention (i.e. alone or in combination with radiation and/or chemotherapeutic agents) may be prepared in the form of its salts (particularly its pharmaceutically acceptable salts), N-oxides, hydrates, solvates and polymorphic forms thereof.
6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide may be administered in a variety of dosage forms. Thus, it can be administered orally, for example as tablets, capsules, pastilles (troche), lozenges (lozenge), aqueous or oily suspensions, dispersible powders or granules. 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide may be administered in a sublingual formulation (e.g., an oral formulation). 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide may also be administered parenterally, whether subcutaneously, intravenously, intramuscularly, intrasternally, transdermally, by inhalation, intranasally or by infusion techniques. Thus, 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide is administered orally, or by inhalation, or intranasally, but preferably the route of administration is oral or intravenous. In the case of oral administration of 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridin-2-yl ], the preferred carrier is a tablet or capsule. In the latter case, it is generally preferred to administer the compound in the form of a hard gelatin capsule or one of a variety of sustained release formulations known in the art. Where the route of administration is intravenous, 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide is administered as an aqueous solution.
6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide is typically formulated for administration with a pharmaceutically acceptable carrier or diluent. For example, the solid oral forms may contain, in addition to the active compound, diluents, such as lactose, dextrose, sucrose, cellulose, corn starch or potato starch; lubricants, such as silica, talc, stearic acid, magnesium or calcium stearate, and/or polyethylene glycol; an adhesive; such as starch, gum arabic, gelatin, methylcellulose, carboxymethylcellulose, or polyvinylpyrrolidone; disintegrating agents, such as starch, alginic acid, alginates or sodium starch glycolate; foaming the mixture; a dye; a sweetener; wetting agents, such as lecithin, polysorbate, lauryl sulfate; and, in general, nontoxic and pharmacologically inactive substances used in pharmaceutical formulations. Such pharmaceutical preparations may be manufactured in a known manner, for example, by means of mixing, granulating, tabletting, sugar-coating or film-coating processes.
Liquid dispersions for oral administration may be syrups, emulsions and suspensions. Syrups may contain as carrier, for example, sucrose or sucrose with glycerol and/or mannitol and/or sorbitol. Suspensions and emulsions may contain as carrier, for example, a natural gum, agar, sodium alginate, pectin, methylcellulose, carboxymethylcellulose or polyvinyl alcohol. Suspensions or solutions for intramuscular injections may contain, in addition to the active compound, a pharmaceutically acceptable carrier, for example, sterile water, olive oil, ethyl oleate, glycols such as propylene glycol, and, if desired, a suitable amount of lidocaine hydrochloride.
Solutions for injection or infusion may contain a carrier, for example sterile water, or preferably, they may be in the form of a sterile aqueous isotonic saline solution.
It will be understood that the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination and the severity of the particular disease undergoing therapy. The optimal dose level and frequency of administration will be determined by clinical trials as required in the art. However, it is expected that a typical dose of 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide will be in the range of about 0.001 to 50mg/kg body weight.
Synthesis of
6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide may be prepared using techniques known to the skilled artisan, including, for example, the procedures shown in scheme 1.
Figure BDA0002833537670000141
Scheme 1.6 synthetic route for- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide
The following abbreviations have been used:
Figure BDA0002833537670000142
Figure BDA0002833537670000151
experimental methods
Unless otherwise stated, all reagents were of commercial grade and used as received without further purification. Unless otherwise indicated, reagent grade solvents were used. Reactions promoted by microwave heating were performed on a Biotage Initiator system. Preparative low pressure chromatography was performed using a CombiFlash company or CombiFlash RF system equipped with RediSep or grace resolution silica and a C18 reverse phase chromatography column. Preparative reverse phase HPLC was performed on a Gilson system with a UV detector equipped with an ACE-5AQ, 100X 21.20mm, 5mm or Phenomenex Synergi Hydro-RP 80A AXIA, 100X 21.20mm, 4mm column. The purest fractions were collected, concentrated and dried under vacuum. The compounds are usually dried in a vacuum oven at between 40 ℃ and 60 ℃ and then analyzed for purity. Analytical HPLC was performed on an Agilent 1100 system. Analytical LCMS was performed on an Agilent 1100HPLC system using a Waters ZQ mass spectrometer. NMR was performed on Bruker Avance 500MHz Cryo Ultrashield with a Dual cryotherapy probe (Dual cryoProbe). IR analysis was performed on a Perkin Elmer FT-IR Spectrum BX using Pike MIRacle single reflection ATR. Melting point determinations were performed on a Reichert Thermovar high temperature hot stage microscope (hotspot microscope). Unless otherwise stated, the reaction is carried out at room temperature. Compounds were automatically named using IUPAC rules.
Intermediate 1
6-chloro-N- (pyridin-4-yl) pyridine-2-carboxamide
6-chloropyridine-2-carboxylic acid (5.50g, 34.9mm0l) and DMF (0.5mL) were dissolved in DCM (100mL) and oxalyl chloride (7.09mL, 83.8mmol) was added. The reaction mixture was stirred for 0.5 h, then the solvent was removed in vacuo. The residue was dissolved in DCM (100mL) cooled to 0 ℃. DIPEA (14.6mL, 83.8mmol) and 4-aminopyridine (3.94g, 41.9mm0l) were added and the reaction was warmed to room temperature and then stirred for an additional 0.5 h. The solvent was removed in vacuo and the residue partitioned between DCM (100mL) and water (75 mL). The aqueous layer was extracted with DCM (2X 75mL), the organic layers were combined and Na was added2CO3(1M, 75mL), brine (75mL), and dried (MgSO)4) And the solvent was removed in vacuo. The residue was purified by column chromatography to give the title compound as an off-white solid (6.66g, 81.7%). LCMS (ES)+):234.2[MH]+
Intermediate 2
6- (1, 4-diazepan-1-yl) -N- (pyridin-4-yl) pyridine-2-carboxamide
Intermediate 1(1.5g, 6.42mmol) was dissolved in DMA (12.5 mL). Homopiperazine (3.22g, 32.1mmol) was added and the reaction mixture was heated at 180 ℃ for 0.5 h using a Biotage microwave. This process was repeated three more times on the same scale and the four batches were combined and the solvent removed in vacuo. The residue was dissolved in DCM (300mL) and saturated Na2CO3Washed with aqueous solution (150mL), brine (100mL), and dried (MgSO)4) And the solvent was removed in vacuo. The residue was purified by column chromatography to give the title compound (6.88g, 90.1%) as a pale yellow solid. LCMS (ES)+):298.2[MH]+
6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide
Intermediate 2(4.88g, 16.4mmol) was dissolved in DCM (200 mL). 1- (Prop-2-yl) piperidin-4-one (4.88mL, 32.8mmol) and sodium triacetoxyborohydride (17.4g, 82.1mmol) were added and the reaction mixture was stirred for 20 hours. The reaction mixture was diluted with DCM (200mL) and saturated Na2CO3Aqueous solution (100mL) was quenched. The aqueous layer was extracted with DCM (100 mL). The combined organic layers were washed with brine (50mL) and dried (MgSO)4),And the solvent was removed in vacuo. The residue was purified by crystallization from MeCN followed by reverse phase column chromatography. The residue was taken up in DCM (300mL) and saturated Na2CO3The aqueous solution (100mL) was partitioned. The aqueous layer was extracted with DCM (50mL), the organic layers were combined, washed with brine (50mL), dried (MgSO)4) And the solvent was removed in vacuo. The residue was crystallized from MeCN to give the title compound as a pale yellow solid (4.66g, 67.3%).
HPLC: room temperature 3.47 min, 100% purity
LCMS(ES+):423.2[MH]+
1H NMR(500 MHz,DMSO-d6H 10.31(1H,s,NH),8.52-8.50(2H,m,ArH),7.84-7.82(2H,m,ArH),7.70(1H,dd,J 8.5and 7.3Hz,ArH),7.30(1H,d,J 7.2Hz,ArH),6.93(1H,d,J 8.7Hz,ArH),3.80(2H,m,NCH 2),3.76(2H,m,NCH 2),2.82-2.79(2H,m,NCH 2),2.77-2.73(2H,m,NCH 2),2.62(1H,spt,J 6.6Hz,CHMe),2.58-2.56(2H,m,NCH 2),2.39-2.33(1H,m,NCHCH2),2.05-1.88(2H,m,NCH 2),1.85-1.78(2H,m,CH 2),1.65-1.60(2H,m,NCHCH 2),1.36(2H,qd,J 11.7and 3.4Hz,NCHCH 2),0.91(6H,d,J 6.6Hz.CH(CH 3)2)
IR (solid) vMaximum of/cm-13328, 2936, 2358, 2162, 1982, 1682, 1597, 1582, 1510, 1485, 1459, 1418, 1404, 1383, 1364, 1336, 1282, 1246, 1211, 1179, 1161, 1125, 1070, 1030, 994, 972, 926, 898, 878, 824, 814, 758, 681, and 617.
Melting point: 157 ℃ 159 DEG C
The following examples are provided to further illustrate embodiments of the present invention.
Example 1
In the experiment shown in FIG. 1, a group of 5 mice was injected with vehicle AMD3100(5mg/kg) or subcutaneously with 6- {4- [1- (prop-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide (30mg/kg) and the mobilization of hematopoietic progenitor cells was assessed after 1 hour. Data are expressed as colony forming units of pluripotent GEMM cells per mL of peripheral blood.
Figure 1 reveals that 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide does not result in significant mobilization of HSCs from mouse bone marrow. This was unexpected in view of the propensity of known CXCR4 antagonists (e.g., AMD 3100/plerixafor/Mozobil) to mobilize HSCs. Figure 1 shows that the mobilization of HSCs by AMD3100 is significantly higher than that caused by vehicle (P < 0.05). Mobilizing a reduction in HSCs from the protective environment of the bone marrow is expected to reduce the risk of side effects caused by HSC destruction by irradiation and/or chemotherapeutic agents, such side effects including anemia and neutropenia.
Example 2
In this example, the efficacy of 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide in inhibiting the growth of the human glioblastoma cell line (T98G) in subcutaneous xenografts in nude mice was demonstrated (figure 2). FIG. 2 shows the inhibition of T98G xenograft growth in nude mice by 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide (50mg/kg, once daily by oral gavage, 5 days a week, dotted line). The solid line represents the control (i.e., untreated). The data in figure 2 are expressed as a percentage of tumor progression in a group of 8 to 10 mice, where progression is defined as a 20% increase in tumor volume. Tumor volume was determined by measuring the width and depth of the tumor with calipers and then calculating the volume. The x-axis shows days. After 15 days, all control mice had progressed, whereas none of the mice treated with 6- {4- [1- (prop-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide had progressed. Inhibition of the growth of the human glioblastoma cell line (T98G) in nude mice is expected to be predictive of beneficial therapeutic outcomes in human cancer patients, including patients with glioblastoma and astrocytoma.
Example 3
In this example, 6- {4- [1- (propan-2-yl) piperidin-4-yl is demonstrated]-1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide efficacy in inhibiting the growth of human glioblastoma cell line (T98G) in subcutaneous xenografts of nude mice (figure 3). Growth of tumors to at least 120mm under the skin3Thereafter, mice were randomized and treated with temozolomide (16mg/kg per day per oral dose for 5 days) and 6- {4- [1- (propan-2-yl) piperidin-4-yl]-1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide (50mg/kg, by oral gavage once daily, 5 days a week, dotted line).
The data in figure 3 are expressed as% of tumor progression in groups of 8 to 10 mice, where progression is defined as a 20% increase in tumor volume. The x-axis shows days; (-) represents an untreated mouse; (- - - - - - - -) represents temozolomide alone; (. cndot.) represents 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide alone; and (-. cndot. -) represents a combination of 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide and temozolomide.
Treatment with 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide and the brain penetrating chemotherapeutic temozolomide is shown in FIG. 3 to inhibit growth of a human CNS (T98G) in nude mouse subcutaneous xenografts. Combining both treatments results in unexpectedly improved (i.e., synergistic) antitumor efficacy. This combination has an advantageously reduced risk of side effects due to the unexpectedly low tendency of 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide to release HSCs from a protective bone marrow environment.
Examples 4 to 10
Introduction to
The efficacy of 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide alone or in combination with bevacizumab, temozolomide, radiation therapy or sunitinib in inhibiting the growth of in situ (intracranial) tumors in nude mice is demonstrated in figures 4-7, 8A, 8B, 9A, 9B and 10. The x-axis shows the number of days in each graph. In the graphs of fig. 4 to 7 and 10, the y-axis shows the% survival (i.e., the% of mice that have not yet been euthanized). In the graphs of fig. 8A, 8B, 9A and 9B, the y-axis shows the percentage of tumor progression, where progression is defined as the point in time when the tumor grows to a size detectable by luminescence. The experiments represented by figures 4, 5 and 6 have the same control (i.e., vehicle only) and therefore the same control data. To improve clarity, the lines representing the control data have been removed from fig. 5 and 6, but remain in fig. 4.
Materials and methods
Examples 4 to 10 use the following materials and methods unless otherwise indicated.
The nude mice were fixed on the stereotaxic apparatus and anesthetized. The surgical area is prepared with povidone iodine (betadine). A small hole was formed 1.0mm in front of and 2mm outside the exposed bregma. A sterile 5gL Hamilton syringe with a 26 gauge needle was inserted from the surface of the skull to a depth of 3.0mm and withdrawn 0.5mm to inject a 3X 10 syringe into a 3 μ L volume3U87MG cells. The injection rate was set at 1 μ L/min. After implantation of the tumor cells, the needle was left in place for 5 minutes to prevent reflux. The needle was then completely removed from the brain over the course of 4 minutes (1.0 mm/min) and the skin was sutured. Before treatment initiation (5 days after injection), animals were randomly divided into treatment groups of 10 mice. Selection of a small number of cells (3X 10)3) To simulate a chemo-radiation treatment performed after surgery, in which a small amount of tumor cells remain in the operating bed, regrow and produce a recurrence. Treatment was initiated 5 days after cell injection when luciferase activity was not detected intracranial for 35 days. The time to progression (i.e., luminescence detected) was assessed and mice were followed up to 180 days. Mice were euthanized when they exhibited neurological symptoms (e.g., altered gait, tremor/seizure, lethargy) or weight loss of 20% or more of the pre-operative body weight. The y-axis parameter "survival" is the percentage of mice that have not yet been euthanized. The y-axis parameter "probability of detection" is of mice with tumors that have progressed to the stage at which luminescence is detectedPercentage (D).
The following doses were used: 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide was administered orally at 50mg/kg once daily. Bevacizumab was administered intravenously at 4mg/kg every 4 days. Temozolomide was administered orally at 32mg/kg daily. Sunitinib was administered orally at 40mg/kg daily. Radiation therapy consisted of 3 × 2Gy per day.
Results and conclusions
An increase in survival of nude mice with intracranial tumors formed by a human glioblastoma cell line (e.g., U87MG) is expected to be predictive of beneficial therapeutic outcomes in human cancer patients, including patients with CNS cancers such as glioblastoma and astrocytoma.
An increase in the time required for an intracranial tumor formed by a human glioblastoma cell line (e.g., U87MG) detectable by luminescence in nude mice is expected to be predictive of beneficial therapeutic outcomes in human cancer patients, including patients with CNS cancers such as glioblastomas and astrocytomas.
Referring now to the drawings, figure 4 shows that 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide acts synergistically with bevacizumab in increasing survival of mice with in situ (intracranial) tumors (in combination p ═ 0.002, HR 3.4 relative to vehicle).
Figure 5 shows that 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide acts synergistically with temozolomide in increasing survival of mice with orthotopic (intracranial) tumors (p ═ 0.02 in combination, HR 2.8 versus temozolomide alone).
Figure 6 shows that 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide acts synergistically with radiotherapy in increasing survival of mice with orthotopic (intracranial) tumors (combination p ═ 0.0002, HR 4.0 versus radiotherapy alone).
Figure 7 shows that 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide synergistically enhances the effect of sunitinib in increasing survival of mice with orthotopic (intracranial) tumors (combination p ═ 0.2, HR 1.6 versus vehicle).
As shown in figures 8A and 8B, 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide acts synergistically with bevacizumab or sunitinib in increasing the time required for tumor progression, demonstrating inhibition of tumor growth and increased probability of survival (figure 8A; combined p ═ 0.0001, HR 9.7 versus vehicle) and with sunitinib (figure B; combined p ═ 0.0001, HR 5.3 versus vehicle). 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide delays the process of tumor growth to a size detectable by luminescence (HR 3.5 vs. vehicle). The combination of 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide with bevacizumab or sunitinib showed significantly improved growth retardation compared to 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide with bevacizumab or sunitinib alone. The Y-axis in fig. 8A and 8B is the same, i.e.: probability (%) of detection.
As shown in fig. 9A and 9B, 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide works synergistically with irradiation and/or temozolomide in the treatment of intracranial tumors. Administration of 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide alone delayed tumor growth (HR 3.7, p < 0.001) and unexpectedly improved the efficacy of irradiation treatment (fig. 9A; irradiation p ═ 0001, HR 4.6 vs. combination) and temozolomide treatment (fig. 9B; temozolomide p ═ 0.01, HR 2.9 vs. combination). The Y-axis in fig. 9A and 9B is the same, i.e.: probability (%) of detection.
As shown in FIG. 10, the 6- {4- [1- (propan-2-yl) piperidin-4-yl group was used]Treatment of-1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide synergizes with respect to survival of mice with in situ (intracranial) tumors with irradiation therapy and temozolomide therapy (combination p ═ 0).025, HR 2.3). For this experiment, 5 × 10 was injected using the technique described above3U87MG cells. After injection of U87MG cells, tumors were detectable in mice treated for 28 days.

Claims (23)

  1. 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide, or a pharmaceutically acceptable salt thereof, for use in the treatment of CNS cancer.
  2. Use of 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of CNS cancer.
  3. 3. A method of treating a patient having a CNS cancer, said method comprising administering 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide, or a pharmaceutically acceptable salt thereof, to said patient in an amount sufficient to provide a therapeutic effect.
  4. 4. A compound, use or method as claimed in any one of claims 1 to 3 wherein the CNS cancer is brain cancer.
  5. 5. The compound, use or method according to any one of claims 1 to 4, wherein the CNS cancer is glioma.
  6. 6. The compound, use or method according to any one of claims 1 to 5, wherein said CNS cancer is selected from: neuroblastoma, glioblastoma, other astrocytoma, oligodendroglioma, meningioma, ependymoma, oligodendroglioma, medulloblastoma and metastatic cancer that enters the CNS from peripheral cancers.
  7. 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide, or a pharmaceutically acceptable salt thereof, in combination with radiation and/or chemotherapeutic agents for use in the treatment of cancer.
  8. Use of 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in combination with radiation and/or a chemotherapeutic agent in the treatment of cancer.
  9. 9. A method of treating a patient suffering from cancer, said method comprising administering to the patient a therapeutically effective amount of 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide, or a pharmaceutically acceptable salt thereof, in combination with an effective amount of radiation and/or a chemotherapeutic agent in an amount sufficient to provide a therapeutic effect.
  10. 10. A compound, use or method according to any one of claims 7 to 9 wherein the cancer includes the following cancers and metastases thereof: cancers of the lung (including non-small cells and small cells), pancreas, cervix, thyroid, kidney, ovary, prostate, skin (including melanoma); cancers of the GI tract (including esophagus, liver, colorectal, and stomach); squamous cell carcinoma of the oral cavity; blood cancers including leukemias such as B-CLL, AML, CML, ALL; lymphomas such as intraocular, non-hodgkin and hodgkin lymphomas, and multiple myeloma; cancers of the nervous system, including brain cancer, neuroblastoma, glioblastoma, other astrocytoma, oligodendroglioma, meningioma, ependymoma, oligodendroglioma, medulloblastoma, and metastatic cancers that enter the CNS from peripheral cancers.
  11. 11. A compound, use or method according to any one of claims 7 to 9 wherein the cancer is a CNS cancer selected from: neuroblastoma, glioblastoma, other astrocytoma, oligodendroglioma, meningioma, ependymoma, oligodendroglioma, medulloblastoma and metastatic cancer that enters the CNS from peripheral cancers.
  12. 12. The compound, use or method according to any one of claims 7 to 11 wherein the chemotherapeutic agent is a DNA modifier.
  13. 13. A compound, use or method as claimed in any one of claims 7 to 11 wherein the chemotherapeutic agent is selected from: bevacizumab, sunitinib, temozolomide, vincristine, lomustine, procarbazine, carmustine, irinotecan, cisplatin, carboplatin, methotrexate, etoposide, bleomycin, vinblastine, actinomycin D, cyclophosphamide and ifosfamide.
  14. 14. The compound, use or method according to any one of claims 7 to 13, wherein the chemotherapeutic agent is capable of penetrating and achieving a therapeutic concentration in the brain after systemic administration to a patient.
  15. 15. A compound, use or method as claimed in claim 14 wherein the brain penetrating chemotherapeutic agent is selected from any one of sunitinib, lomustine, temozolomide, carmustine, irinotecan and carboplatin.
  16. 16. A compound, use or method according to claim 15 wherein the brain penetrating chemotherapeutic agent is lomustine or carmustine.
  17. 17. A compound, use or method as claimed in any one of claims 7 to 16 wherein the 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide is administered prior to administration of the radiation and/or chemotherapeutic agent.
  18. 18. A compound, use or method as claimed in any one of claims 7 to 16 wherein the 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide is administered simultaneously with the administration of the radiation and/or chemotherapeutic agent.
  19. 19. A compound, use or method as claimed in any one of claims 7 to 16 wherein the 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl } -N- (pyridin-4-yl) pyridine-2-carboxamide is administered after administration of the radiation and/or chemotherapeutic agent.
  20. 20. A compound, use or method as claimed in any one of claims 1 to 19 wherein the cancer to be treated comprises a tumour that is resistant to temozolomide and/or irradiation.
  21. 21. A compound, use or method as claimed in any one of claims 1 to 20 wherein the 6- {4- [1- (propan-2-yl) piperidin-4-yl ] -1, 4-diazepan-1-yl ] -N- (pyridin-4-yl) pyridine-2-carboxamide is in an intravenous formulation.
  22. 22. A compound, use or method as claimed in any one of claims 7 to 21 wherein the chemotherapeutic agent is in an intravenous formulation.
  23. 23. A compound, use or method according to any one of claims 7 to 22 wherein the chemotherapeutic agent is bevacizumab or sunitinib.
CN202011468330.5A 2015-04-02 2016-04-01 Novel treatment for cancer Pending CN112569242A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201562141980P 2015-04-02 2015-04-02
US62/141,980 2015-04-02
CN201680020617.7A CN107438437B (en) 2015-04-02 2016-04-01 For the treatment of cancer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CN201680020617.7A Division CN107438437B (en) 2015-04-02 2016-04-01 For the treatment of cancer

Publications (1)

Publication Number Publication Date
CN112569242A true CN112569242A (en) 2021-03-30

Family

ID=55702039

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202011468330.5A Pending CN112569242A (en) 2015-04-02 2016-04-01 Novel treatment for cancer
CN201680020617.7A Expired - Fee Related CN107438437B (en) 2015-04-02 2016-04-01 For the treatment of cancer

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN201680020617.7A Expired - Fee Related CN107438437B (en) 2015-04-02 2016-04-01 For the treatment of cancer

Country Status (19)

Country Link
US (1) US11311552B2 (en)
EP (1) EP3277284B1 (en)
JP (1) JP6927882B2 (en)
KR (1) KR20170132314A (en)
CN (2) CN112569242A (en)
AU (1) AU2016242118B2 (en)
BR (1) BR112017019437A2 (en)
CA (1) CA2979167A1 (en)
DK (1) DK3277284T3 (en)
EA (1) EA037555B1 (en)
ES (1) ES2826558T3 (en)
HK (1) HK1245120A1 (en)
HU (1) HUE051341T2 (en)
IL (1) IL254149A0 (en)
MX (1) MX2017011981A (en)
PL (1) PL3277284T3 (en)
PT (1) PT3277284T (en)
SG (2) SG11201707429VA (en)
WO (1) WO2016157149A1 (en)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201017345D0 (en) 2010-10-14 2010-11-24 Proximagen Ltd Receptor antagonists
CN112569242A (en) 2015-04-02 2021-03-30 普罗克斯马根有限责任公司 Novel treatment for cancer
GB201604213D0 (en) 2016-03-11 2016-04-27 Proximagen Ltd Drug combination and its use in therapy
GB201703907D0 (en) * 2017-03-10 2017-04-26 Proximagen Ltd Novel therapies for cancer
WO2020222189A1 (en) 2019-05-02 2020-11-05 Proximagen, Llc Crystalline form of 6-[4-[1-(propan-2-yl)piperidin-4-yl]-1,4-diazepan-1-yl]-n-(pyrdin-4-yl)pyridine-2-carboxamide
WO2020222190A1 (en) 2019-05-02 2020-11-05 Proximagen, Llc Crystalline form of 6-[4-[1 -(propan-2-yl)piperidin-4-yl]-1,4-diazepan-1 -yl]-n-(pyrdin-4-yl)pyridine-2-carboxamide
WO2021261601A1 (en) 2020-06-26 2021-12-30 ラクオリア創薬株式会社 Method for selecting cancer patient for which combination therapy of retinoid and cancer treatment agent will be effective, and combined drug of retinoid and cancer treatment agent

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1679562A (en) * 1998-07-08 2005-10-12 阿诺麦德股份有限公司 Antiviral compounds
CN103282360A (en) * 2010-10-14 2013-09-04 普罗克斯马根有限公司 Cxcr4 receptor antagonists

Family Cites Families (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5047406A (en) 1989-12-06 1991-09-10 Warner-Lambert Co. Substituted cyclohexanols as central nervous system agents
GB9300194D0 (en) 1993-01-06 1993-03-03 Wyeth John & Brother Ltd Piperazine derivatives
US5939098A (en) * 1996-09-19 1999-08-17 Schering Corporation Cancer treatment with temozolomide
WO1998025617A1 (en) 1996-12-13 1998-06-18 Merck & Co., Inc. Substituted aryl piperazines as modulators of chemokine receptor activity
FR2769913B1 (en) 1997-10-16 2000-03-10 Pf Medicament NEW CYCLOHEXANIC DERIVATIVES DIFUNCTIONALIZED IN 1, 4, THEIR PREPARATION AND THEIR HUMAN THERAPEUTIC APPLICATION
SE9902987D0 (en) 1999-08-24 1999-08-24 Astra Pharma Prod Novel compounds
AU6871801A (en) 2000-06-29 2002-01-14 Abbott Lab Aryl phenylheterocyclyl sulfide derivatives and their use as cell adhesion-inhibiting anti-inflammatory and immune-suppressive agents
AR036366A1 (en) 2001-08-29 2004-09-01 Schering Corp USEFUL PIPERIDINE DERIVATIVES AS CCR5 ANTAGONISTS, PHARMACEUTICAL COMPOSITIONS, THE USE OF SUCH DERIVATIVES FOR THE MANUFACTURE OF A MEDICINAL PRODUCT AND A KIT
AU2003220968A1 (en) 2002-03-28 2003-10-13 Kyowa Hakko Kogyo Co., Ltd. Anti-inflammatory agent
US7759336B2 (en) 2002-12-10 2010-07-20 Ono Pharmaceutical Co., Ltd. Nitrogen-containing heterocyclic compounds and medicinal use thereof
CA2520406A1 (en) * 2003-03-27 2004-10-14 Emory University Cxcr4 antagonists and methods of their use
GB0308466D0 (en) 2003-04-11 2003-05-21 Novartis Ag Organic compounds
FR2854633B1 (en) 2003-05-07 2005-06-24 Sanofi Synthelabo PIPERIDINYL-AND PIPERAZINYL-ALKYLCARBAMATES DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC USE
US7498346B2 (en) 2003-12-11 2009-03-03 Genzyme Corporation Chemokine receptor binding compounds
WO2005059107A2 (en) 2003-12-11 2005-06-30 Anormed Inc. Chemokine receptor binding compounds
US7880002B2 (en) 2004-12-29 2011-02-01 Millennium Pharmaceuticals, Inc. Substituted piperazinyl-pyrrolidine compounds useful as chemokine receptor antagonists
US7635698B2 (en) 2004-12-29 2009-12-22 Millennium Pharmaceuticals, Inc. Compounds useful as chemokine receptor antagonists
WO2006088921A2 (en) 2005-02-16 2006-08-24 Schering Corporation Pyrazinyl substituted piperazine-piperidines with cxcr3 antagonist activity
EP1856097B1 (en) 2005-02-16 2012-07-11 Schering Corporation Pyridyl and phenyl substituted piperazine-piperidines with cxcr3 antagonist activity
WO2006088840A1 (en) 2005-02-16 2006-08-24 Schering Corporation Novel heterocyclic substituted pyridine or phenyl compounds with cxcr3 antagonist activity
CN101189238A (en) 2005-02-16 2008-05-28 先灵公司 Piperazine-piperidines with CXCR3 antagonist activity
WO2006088920A1 (en) 2005-02-16 2006-08-24 Schering Corporation Amine-linked pyridyl and phenyl substituted piperazine-piperidines with cxcr3 antagonist activity
EP2527337A1 (en) 2005-04-14 2012-11-28 Bristol-Myers Squibb Company Inhibitors of 11-beta hydroxysteroid dehydrogenase type I
WO2006130426A2 (en) 2005-05-27 2006-12-07 Kemia, Inc. Modulators of ccr-5 activity
GB0525957D0 (en) 2005-12-21 2006-02-01 Astrazeneca Ab Methods
CN101460482B (en) 2006-03-21 2013-03-27 默沙东公司 Heterocyclic substituted pyridine compounds with cxcr3 antagonist activity
EP1849781A1 (en) 2006-04-28 2007-10-31 Laboratorios del Dr. Esteve S.A. Substituted 3-Amino-4-hydroxy pyrrolidines compounds, their preparation and use as medicaments
AR061975A1 (en) 2006-07-14 2008-08-10 Schering Corp PIPERAZINE COMPOUNDS 1,4-REPLACED WITH ANTIGONIST ACTIVITY OF CXCR3, A PHARMACEUTICAL COMPOSITION THAT INCLUDES THEM AND ITS USE IN THE TREATMENT OF DISEASES MEDIATED BY THE CXCR3 CHEMIOQUINE RECEIVER
JP5606736B2 (en) 2006-09-02 2014-10-15 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Particle beam method for orienting reactive mesogens.
NZ575907A (en) 2006-09-21 2011-03-31 Kyorin Seiyaku Kk 5-substituted benzoxazines
MX2009005252A (en) 2006-11-17 2009-05-28 Abbott Lab Aminopyrrolidines as chemokine receptor antagonists.
US20120157466A1 (en) 2006-12-22 2012-06-21 Qingbei Zeng Heterocyclic compounds with cxcr3 antagonist activity
NZ578744A (en) 2007-01-31 2012-02-24 Vertex Pharma 2-aminopyridine derivatives useful as kinase inhibitors
WO2008121065A1 (en) 2007-03-30 2008-10-09 Astrazeneca Ab Novel pyrrolidine derivatives as antagonists of the chemokine receptor
BRPI0821151A2 (en) 2007-12-07 2015-06-16 Novartis Ag Pyrazole derivatives and their use as cyclin-dependent kinase inhibitors
WO2009108360A2 (en) * 2008-02-29 2009-09-03 The Board Of Trustees Of The Leland Stanford Junior University Inhibition of post-radiation tumor growth
JP5785089B2 (en) 2008-11-04 2015-09-24 ケモセントリックス,インコーポレイティド Regulators of CXCR7
EP2454257B1 (en) 2009-07-15 2013-08-21 AbbVie Inc. Pyrrolopyridine inhibitors of kinases
KR20110123657A (en) 2010-05-07 2011-11-15 에스케이케미칼주식회사 Picolinamide and pyrimidine-4-carboxamide compounds, process for preparing and pharmaceutical composition comprising the same
WO2012047339A2 (en) 2010-06-28 2012-04-12 The General Hospital Corporation Anti-cxcr4 as a sensitizer to cancer therapeutics
GB201017048D0 (en) * 2010-10-08 2010-11-24 Ucl Business Plc Composition
EP2935215B1 (en) * 2012-12-20 2019-11-13 Lu, Qing-Bin Radiosensitizer compounds for use in combination with radiation
CN112569242A (en) 2015-04-02 2021-03-30 普罗克斯马根有限责任公司 Novel treatment for cancer
GB201604213D0 (en) 2016-03-11 2016-04-27 Proximagen Ltd Drug combination and its use in therapy
GB201703907D0 (en) 2017-03-10 2017-04-26 Proximagen Ltd Novel therapies for cancer

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1679562A (en) * 1998-07-08 2005-10-12 阿诺麦德股份有限公司 Antiviral compounds
CN103282360A (en) * 2010-10-14 2013-09-04 普罗克斯马根有限公司 Cxcr4 receptor antagonists

Also Published As

Publication number Publication date
AU2016242118B2 (en) 2021-07-08
EA037555B1 (en) 2021-04-13
EA201791953A1 (en) 2018-01-31
PL3277284T3 (en) 2021-03-08
EP3277284A1 (en) 2018-02-07
WO2016157149A9 (en) 2016-12-08
ES2826558T3 (en) 2021-05-18
BR112017019437A2 (en) 2018-05-02
PT3277284T (en) 2020-11-11
WO2016157149A1 (en) 2016-10-06
HK1245120A1 (en) 2018-08-24
US11311552B2 (en) 2022-04-26
MX2017011981A (en) 2018-01-10
SG11201707429VA (en) 2017-10-30
CA2979167A1 (en) 2016-10-06
DK3277284T3 (en) 2020-10-26
AU2016242118A1 (en) 2017-10-05
US20180078563A1 (en) 2018-03-22
CN107438437A (en) 2017-12-05
JP2018510154A (en) 2018-04-12
HUE051341T2 (en) 2021-03-01
IL254149A0 (en) 2017-10-31
JP6927882B2 (en) 2021-09-01
EP3277284B1 (en) 2020-08-05
SG10201909188XA (en) 2019-11-28
KR20170132314A (en) 2017-12-01
CN107438437B (en) 2021-01-01

Similar Documents

Publication Publication Date Title
CN107438437B (en) For the treatment of cancer
US11833147B2 (en) Procaspase 3 activation by combination therapy
US20100129470A1 (en) Method of treating brain cancer
TW201217361A (en) Method of treating abnormal cell growth
CN109641009B (en) Antitumor agent, antitumor effect enhancer, and antitumor kit
WO2019176985A1 (en) Antitumor agent, antitumor effect potentiator and antitumor kit
AU2013230994B2 (en) Procaspase 3 activation by combination therapy
TW202339748A (en) Treatment methods with substituted pyrimidin-4(3h)-ones
JP2023539715A (en) Combination of antibody-drug conjugates and ATM inhibitors
KR20200061817A (en) Use of pyrazolone derivatives having apoptosis of brain cancer stem cells
KR20220007146A (en) Quinoline Derivatives Used for Combination Treatment of Soft Tissue Sarcoma

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20210330

WD01 Invention patent application deemed withdrawn after publication